1
|
Tran MH, Gao J, Wang X, Liu R, Parris CL, Esquivel C, Fan Y, Wang L. Enhancing Liver Transplant Outcomes through Liver Precooling to Mitigate Inflammatory Response and Protect Mitochondrial Function. Biomedicines 2024; 12:1475. [PMID: 39062048 PMCID: PMC11275024 DOI: 10.3390/biomedicines12071475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Transplanted organs experience several episodes of ischemia and ischemia-reperfusion. The graft injury resulting from ischemia-reperfusion (IRI) remains a significant obstacle to the successful survival of transplanted grafts. Temperature significantly influences cellular metabolic rates because biochemical reactions are highly sensitive to temperature changes. Consequently, lowering the temperature could reduce the degradative reactions triggered by ischemia. In mitigating IRI in liver grafts, the potential protective effect of localized hypothermia on the liver prior to blood flow obstruction has yet to be explored. In this study, we applied local hypothermia to mouse donor livers for a specific duration before stopping blood flow to liver lobes, a procedure called "liver precooling". Mouse donor liver temperature in control groups was controlled at 37 °C. Subsequently, the liver donors were preserved in cold University of Wisconsin solution for various durations followed by orthotopic liver transplantation. Liver graft injury, function and inflammation were assessed at 1 and 2 days post-transplantation. Liver precooling exhibited a significant improvement in graft function, revealing more than a 47% decrease in plasma aspartate transaminase (AST) and alanine aminotransferase (ALT) levels, coupled with a remarkable reduction of approximately 50% in liver graft histological damage compared to the control group. The protective effects of liver precooling were associated with the preservation of mitochondrial function, a substantial reduction in hepatocyte cell death, and a significantly attenuated inflammatory response. Taken together, reducing the cellular metabolism and enzymatic activity to a minimum level before ischemia protects against IRI during transplantation.
Collapse
Affiliation(s)
- Minh H. Tran
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Jie Gao
- School of Health Professions, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xinzhe Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Colby L. Parris
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Carlos Esquivel
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Yingxiang Fan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
2
|
NOX as a Therapeutic Target in Liver Disease. Antioxidants (Basel) 2022; 11:antiox11102038. [PMID: 36290761 PMCID: PMC9598239 DOI: 10.3390/antiox11102038] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
The nicotinamide adenine dinucleotide phosphate hydrogen oxidase (NADPH oxidase or NOX) plays a critical role in the inflammatory response and fibrosis in several organs such as the lungs, pancreas, kidney, liver, and heart. In the liver, NOXs contribute, through the generation of reactive oxygen species (ROS), to hepatic fibrosis by acting through multiple pathways, including hepatic stellate cell activation, proliferation, survival, and migration of hepatic stellate cells; hepatocyte apoptosis, enhancement of fibrogenic mediators, and mediation of an inflammatory cascade in both Kupffer cells and hepatic stellate cells. ROS are overwhelmingly produced during malignant transformation and hepatic carcinogenesis (HCC), creating an oxidative microenvironment that can cause different and various types of cellular stress, including DNA damage, ER stress, cell death of damaged hepatocytes, and oxidative stress. NOX1, NOX2, and NOX4, members of the NADPH oxidase family, have been linked to the production of ROS in the liver. This review will analyze some diseases related to an increase in oxidative stress and its relationship with the NOX family, as well as discuss some therapies proposed to slow down or control the disease's progression.
Collapse
|
3
|
Hines IN, Milton J, Kremer M, Wheeler MD. Ablation of Tumor Necrosis Factor Alpha Receptor 1 Signaling Blunts Steatohepatitis in Peroxisome Proliferator Activated Receptor α-Deficient Mice. MEDICAL RESEARCH ARCHIVES 2022; 10:3082. [PMID: 36865784 PMCID: PMC9977327 DOI: 10.18103/mra.v10i9.3082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Tumor necrosis factor -alpha (TNFα) is strongly associated with fatty liver disease (i.e, hepatosteatosis). Cytokine production has been thought of as a consequence of hepatic lipid accumulation which becomes a critical factor in the development of chronic liver pathologies as well as insulin resistance. The purpose of this study was to test the hypothesis that TNFα directly regulates lipid metabolism in liver in the mutant peroxisome-proliferator activated receptor-alpha (PPARα-/-) mouse model with robust hepatic lipid accumulation. At 10 weeks of age, TNFα and TNF receptor 1 expression are increased in livers of PPARα-/- mice compared to wild type. PPARα-/- mice were then crossed with mice lacking the receptor for TNFα receptor 1 (TNFR1-/-). Wild type, PPARα-/-, TNFR1-/-, PPARα-/- x TNFR1-/- mice were housed on ad-libitum standard chow diet for up to 40 weeks. Increases in hepatic lipid and liver injury and metabolic disruption associated with PPARα ablation were largely blunted when PPARα-/- mice were crossed with TNFR1-/- mice. These data support the hypothesis that TNFR1 signaling is critical for accumulation of lipid in liver. Therapies that reduce pro-inflammatory responses, namely TNFα, could have important clinical implications to reduce hepatosteatosis and progression of severe liver disease.
Collapse
Affiliation(s)
- Ian N. Hines
- Department of Nutrition Science, East Carolina University, North Carolina, USA
| | - Jamie Milton
- Department of Nutrition Science, East Carolina University, North Carolina, USA
| | - Michael Kremer
- Department of General and Visceral Surgery, Hospital of Aarau, Aarau, Switzerland
| | - Michael D. Wheeler
- Department of Nutrition Science, East Carolina University, North Carolina, USA,
| |
Collapse
|
4
|
Xiao Q, Liu Y, Zhang X, Liu Z, Xiao J, Ye Q, Fu B. Mild hypothermia ameliorates hepatic ischemia reperfusion injury by inducing RBM3 expression. Apoptosis 2022; 27:899-912. [PMID: 35930183 DOI: 10.1007/s10495-022-01757-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 11/02/2022]
Abstract
Liver ischemia reperfusion injury (IRI) is a serious complication of certain liver surgeries, and it is difficult to prevent. As a potential drug-free treatment, mild hypothermia has been shown to promote positive outcomes in patients with IRI. However, the protective mechanism remains unclear. We established in vivo and in vitro models of hepatic ischemia reperfusion (IR) and mild hypothermia pretreatment. Hepatocytes were transfected with RNA-binding motif protein 3 (RBM3) overexpression plasmids, and IR was performed. Cell, culture medium, blood and tissue samples were collected to assess hepatic injury, oxidative stress, apoptosis and changes in RBM3 expression in the liver. Upregulation of RBM3 expression by mild hypothermia reduced the aminotransferase release, liver tissue injury and mitochondrial injury induced by liver IR. Hepatic IR-induced p38 and c-Jun N-terminal kinase (JNK) signaling pathway activation, oxidative stress injury and apoptosis could be greatly reversed by mild hypothermia. Overexpression of RBM3 mimicked the hepatoprotective effect of mild hypothermia. Mild hypothermia protects the liver from ischemia reperfusion-induced p38 and JNK signaling pathway activation, oxidative stress injury and apoptosis through the upregulation of RBM3 expression.
Collapse
Affiliation(s)
- Qi Xiao
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yuan Liu
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - XingJian Zhang
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - ZhongZhong Liu
- Institute of Hepatobiliary Diseases, Transplant Center, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - JianSheng Xiao
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - QiFa Ye
- Institute of Hepatobiliary Diseases, Transplant Center, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - BiQi Fu
- Department of Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
An W, Kang JS. Effect of Metformin on Myocardial Injury Induced by Hepatic Ischemia-Reperfusion in Rats. Front Pharmacol 2022; 13:822743. [PMID: 35431970 PMCID: PMC9010783 DOI: 10.3389/fphar.2022.822743] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/11/2022] [Indexed: 01/30/2023] Open
Abstract
Background: There is no effective medication for treatment or prevention of hepatic ischemia-reperfusion (HIR) injury caused by liver transplantation and hepatectomy. This study aimed to investigate the therapeutic effects of metformin on HIR injury and related myocardial injury in rats.Methods: Wistar male rats were randomly divided into four groups: sham group, ischemia-reperfusion group, and IR group treated with metformin 150 mg/kg and 100 mg/kg. Wistar male rats were administered metformin 150 mg/kg, 100 mg/kg or saline 30 min pre-operative and underwent 15 min ischemia and 6 h reperfusion (n = 4).Results: Metformin significantly alleviates the injury caused by HIR. Administration of metformin resulted in a significant reduction in the serum levels of alanine transaminase and aspartate transaminase and the activity of malondialdehyde, creatine kinase-MB, and lactate dehydrogenase but maintained high catalase and superoxide dismutase activity. Metformin significantly inhibited the IR-induced elevation of tumor necrosis factor-α in liver and heart tissue.Conclusion: Metformin can alleviate hepatic and myocardial injury induced by IR by inhibiting oxidative stress.
Collapse
|
6
|
Czajka P, Przybyłkowski A, Nowak A, Postula M, Wolska M, Mirowska-Guzel D, Czlonkowska A, Eyileten C. Antiplatelet drugs and liver fibrosis. Platelets 2022; 33:219-228. [PMID: 33577391 DOI: 10.1080/09537104.2021.1883574] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Liver fibrosis results from an imbalance between extracellular matrix formation and degradation. The background of liver fibrosis is chronic inflammation and subsequent microcirculation disturbance including microthrombosis. Platelets actively participate in liver fibrosis not only as a part of the clotting system but also by releasing granules containing important mediators. In fact, platelets may play a dual role in the pathophysiology of liver fibrosis as they are able to stimulate regeneration as well as aggravate the destruction of the liver. Recent studies revealed that antiplatelet therapy correlates with inhibition of liver fibrosis. However, liver impairment is associated with extensive coagulation disorders thus the safety of antiplatelet therapy is an area for detailed exploration. In this review, the role of platelets in liver fibrosis and accompanying hemostatic disorders are discussed. Additionally, results of animal and human studies on antiplatelet drugs in liver disorders and their potential therapeutic utility are presented.
Collapse
Affiliation(s)
- Pamela Czajka
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Anna Nowak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Marta Wolska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Anna Czlonkowska
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| |
Collapse
|
7
|
Tan L, Liu L, Yao J, Piao C. miR-145-5p attenuates inflammatory response and apoptosis in myocardial ischemia-reperfusion injury by inhibiting (NADPH) oxidase homolog 1. Exp Anim 2021; 70:311-321. [PMID: 33658472 PMCID: PMC8390312 DOI: 10.1538/expanim.20-0160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a common complication following reperfusion therapy that involves a series of immune or apoptotic reactions. Studies have revealed the potential roles of miRNAs in I/R injury. Herein, we established a myocardial I/R model in rats and a hypoxia/reoxygenation (H/R) model in H9c2 cells and investigated the effect of miR-145-5p on myocardial I/R injury. After 3 h or 24 h of reperfusion, left ventricular end-systolic pressure (LVESP), ejection fraction (EF), and fractional shortening (FS) were obviously decreased, and left ventricular end-diastolic pressure (LVEDP) was increased. Meanwhile, I/R induced an increase in myocardial infarction area. Moreover, a decrease in miR-145-5p and increase in (NADPH) oxidase homolog 1 (NOH-1) were observed following I/R injury. With this in mind, we performed a luciferase reporter assay and demonstrated that miR-145-5p directly bound to NOH-1 3' untranslated region (UTR). Furthermore, miR-145-5p mimics decreased the levels of tumor necrosis factor (TNF)-α, IL-1β, and IL-6 via oxygen and glucose deprivation/reperfusion (OGD/R) stimulation. Upregulation of miR-145-5p increased cell viability and reduced apoptosis accompanied by downregulation of Bax, cleaved caspase-3, cleaved poly(ADP-ribose) polymerase (PARP) and upregulation of Bcl2. In addition, miR-145-5p overexpression increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) and malondialdehyde (MDA) content under OGD/R stress. Notably, NOH-1 could significantly abrogate the above effects, suggesting that it is involved in miR-145-5p-regulated I/R injury. In summary, our findings indicated that miR-145-5p/NOH-1 has a protective effect on myocardial I/R injury by inhibiting the inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Lili Tan
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Limin Liu
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Jian Yao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Chenghao Piao
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| |
Collapse
|
8
|
Abstract
Nearly 100 years ago, Otto Warburg undertook a study of tumor metabolism, and discovered increased lactate caused by increased glycolysis in cancer cells. His experiments were conducted in the presence of excess oxygen, but today tumor tissue is known to be a hypoxic environment. However, an increase of glycolysis and lactate production is still a valid observation. Numerous abnormalities and mutations of metabolic enzymes have been found in many cancers. For example, pyruvate kinase M2 has been associated with many cancers and is a major contributor to directing glycolysis into fermentation, forming lactate. Increases in several enzymes, including glucose 6-phosphate dehydrogenase, pyruvate kinase M2, Rad6, or deficiency of other enzymes such as succinate dehydrogenase, all may contribute directly or indirectly to increases in lactate associated with the Warburg effect. In addition, the increased lactate and acid-base changes are modified further by monocarboxylate transporters and carbonic anhydrase, which contribute to alkalinizing tumor cells while acidifying the tumor extracellular environment. This acidification leads to cancer spread. Fully understanding the mechanisms underlying the Warburg effect should provide new approaches to cancer treatment.
Collapse
Affiliation(s)
- Netanya Y Spencer
- Research Division, Joslin Diabetes Center, Boston, MA; Department of Medicine, Harvard Medical School, Boston, MA.
| | - Robert C Stanton
- Research Division, Joslin Diabetes Center, Boston, MA; Department of Medicine, Harvard Medical School, Boston, MA; Nephrology Division, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
9
|
Characteristics of Changes in Inflammatory Cytokines as a Function of Hepatic Ischemia-Reperfusion Injury Stage in Mice. Inflammation 2020; 42:2139-2147. [PMID: 31494794 DOI: 10.1007/s10753-019-01078-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver ischemia-reperfusion injury (IRI) can severely compromise the prognosis of patients receiving liver surgery. While inflammation contributes to the damage resulting from IRI, only a limited number of inflammation biomarkers have been identified as being associated with the different stages of hepatic IRI. As an approach to identify some of these inflammatory cytokines and the molecular mechanisms involved within different stages of hepatic IRI, we used an advanced antibody array assay to detect multiple proteins. With this technology, we observed specific differences in the content of inflammatory cytokines between ischemic and sham controls, as well as a function of the different reperfusion stages in a hepatic IRI mouse model. For example, while liver tissue content of IL-12p40/p70 was significantly increased in the ischemic stage, it was significantly decreased in the reperfusion stage as compared with that of the sham group. For other inflammatory cytokines, no changes were obtained between the ischemic and reperfusion stages with levels of IL-17, Eotaxin-2, Eotaxin, and sTNF RII all being consistently increased, while those of TIMP-1, TIMP-2, BLC, and MCSF consistently decreased as compared with that of the sham group at all reperfusion stages examined. Results from protein function annotation Gene Ontology and the KEGG pathway revealed that inflammatory cytokines are enriched in a network associated with activation of inflammatory response signaling pathways such as TLR, TNF, and IL-17 when comparing responses of the IR versus sham groups. The identification of cytokines along with their roles at specific stages of IRI may reveal important new biological markers for the diagnosis and prognosis of hepatic IRI.
Collapse
|
10
|
Li G, Chan YL, Sukjamnong S, Anwer AG, Vindin H, Padula M, Zakarya R, George J, Oliver BG, Saad S, Chen H. A Mitochondrial Specific Antioxidant Reverses Metabolic Dysfunction and Fatty Liver Induced by Maternal Cigarette Smoke in Mice. Nutrients 2019; 11:nu11071669. [PMID: 31330878 PMCID: PMC6682890 DOI: 10.3390/nu11071669] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/07/2019] [Accepted: 07/18/2019] [Indexed: 11/16/2022] Open
Abstract
Maternal smoking leads to glucose and lipid metabolic disorders and hepatic damage in the offspring, potentially due to mitochondrial oxidative stress. Mitoquinone mesylate (MitoQ) is a mitochondrial targeted antioxidant with high bioavailability. This study aimed to examine the impact of maternal cigarette smoke exposure (SE) on offspring’s metabolic profile and hepatic damage, and whether maternal MitoQ supplementation during gestation can affect these changes. Female Balb/c mice (eight weeks) were either exposed to air or SE for six weeks prior to mating and throughout gestation and lactation. A subset of the SE dams were supplied with MitoQ in the drinking water (500 µmol/L) during gestation and lactation. Intraperitoneal glucose tolerance test was performed in the male offspring at 12 weeks and the livers and plasma were collected at 13 weeks. Maternal SE induced glucose intolerance, hepatic steatosis, mitochondrial oxidative stress and related damage in the adult offspring. Maternal MitoQ supplementation reduced hepatic mitochondrial oxidative stress and improved markers of mitophagy and mitochondrial biogenesis. This may restore hepatic mitochondrial health and was associated with an amelioration of glucose intolerance, hepatic steatosis and pathological changes induced by maternal SE. MitoQ supplementation may potentially prevent metabolic dysfunction and hepatic pathology induced by intrauterine SE.
Collapse
Affiliation(s)
- Gerard Li
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Yik Lung Chan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Suporn Sukjamnong
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Pathum Wan, Bangkok 10330, Thailand
| | - Ayad G Anwer
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Howard Vindin
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Matthew Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Razia Zakarya
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, Sydney, NSW 2037, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
| | - Sonia Saad
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, The University of Sydney, Sydney, NSW 2065, Australia
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia.
- Faculty of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| |
Collapse
|
11
|
Li S, Fujino M, Takahara T, Li XK. Protective role of heme oxygenase-1 in fatty liver ischemia-reperfusion injury. Med Mol Morphol 2019; 52:61-72. [PMID: 30171344 PMCID: PMC6542780 DOI: 10.1007/s00795-018-0205-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022]
Abstract
Ischemia-reperfusion (IR) injury is a kind of injury resulting from the restoration of the blood supply after blood vessel closure during liver transplantation and is the main cause of graft failure. The pathophysiological mechanisms of hepatic IR include a variety of oxidative stress responses. Hepatic IR is characterized by ischemia and hypoxia inducing oxidative stress, immune response and apoptosis. Fat-denatured livers are also used as donors due to the lack of liver donors. Fatty liver is less tolerant to IR than normal liver. Heme oxygenase (HO) is an enzyme that breaks down hemoglobin to bilirubin, ferrous iron and carbon monoxide (CO). Inducible HO subtype HO-1 is an important protective molecule in mammalian cells used to improve acute and chronic liver injury owing to its characteristic anti-inflammatory and anti-apoptotic qualities. HO-1 degrades heme, and its reaction product CO has been shown to reduce hepatic IR injury and increase the survival rate of grafts. As an induced form of HO, HO-1 also exerts a protective effect against liver IR injury and may be useful as a new strategy of ameliorating this kind of damage. This review summarizes the protective effects of HO-1 in liver IR injury, especially in fatty liver.
Collapse
Affiliation(s)
- Shaowei Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Terumi Takahara
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
12
|
von Knethen A, Schäfer A, Kuchler L, Knape T, Christen U, Hintermann E, Fißlthaler B, Schröder K, Brandes RP, Genz B, Abshagen K, Pützer BM, Sha LK, Weigert A, Syed SN, Schulz M, Shah AM, Ernst A, Putyrski M, Finkelmeier F, Pesic M, Greten F, Hogardt M, Kempf VAJ, Gunne S, Parnham MJ, Brüne B. Tolerizing CTL by Sustained Hepatic PD-L1 Expression Provides a New Therapy Approach in Mouse Sepsis. Am J Cancer Res 2019; 9:2003-2016. [PMID: 31037153 PMCID: PMC6485280 DOI: 10.7150/thno.28057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 01/16/2019] [Indexed: 02/06/2023] Open
Abstract
Cytotoxic T lymphocyte (CTL) activation contributes to liver damage during sepsis, but the mechanisms involved are largely unknown. Understanding the underlying principle will permit interference with CTL activation and thus, provide a new therapeutic option. Methods: To elucidate the mechanism leading to CTL activation we used the Hepa1-6 cell line in vitro and the mouse model of in vivo polymicrobial sepsis, following cecal-ligation and -puncture (CLP) in wildtype, myeloid specific NOX-2, global NOX2 and NOX4 knockout mice, and their survival as a final readout. In this in vivo setting, we also determined hepatic mRNA and protein expression as well as clinical parameters of liver damage - aspartate- and alanine amino-transaminases. Hepatocyte specific overexpression of PD-L1 was achieved in vivo by adenoviral infection and transposon-based gene transfer using hydrodynamic injection. Results: We observed downregulation of PD-L1 on hepatocytes in the murine sepsis model. Adenoviral and transposon-based gene transfer to restore PD-L1 expression, significantly improved survival and reduced the release of liver damage, as PD-L1 is a co-receptor that negatively regulates T cell function. Similar protection was observed during pharmacological intervention using recombinant PD-L1-Fc. N-acetylcysteine blocked the downregulation of PD-L1 suggesting the involvement of reactive oxygen species. This was confirmed in vivo, as we observed significant upregulation of PD-L1 expression in NOX4 knockout mice, following sham operation, whereas its expression in global as well as myeloid lineage NOX2 knockout mice was comparable to that in the wild type animals. PD-L1 expression remained high following CLP only in total NOX2 knockouts, resulting in significantly reduced release of liver damage markers. Conclusion: These results suggest that, contrary to common assumption, maintaining PD-L1 expression on hepatocytes improves liver damage and survival of mice during sepsis. We conclude that administering recombinant PD-L1 or inhibiting NOX2 activity might offer a new therapeutic option in sepsis.
Collapse
|
13
|
Malik G, Wilting J, Hess CF, Ramadori G, Malik IA. PECAM-1 modulates liver damage induced by synergistic effects of TNF-α and irradiation. J Cell Mol Med 2019; 23:3336-3344. [PMID: 30761739 PMCID: PMC6484309 DOI: 10.1111/jcmm.14224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 12/29/2022] Open
Abstract
The mechanisms of radiation‐induced liver damage are poorly understood. We investigated if tumour necrosis factor (TNF)‐α acts synergistically with irradiation, and how its activity is influenced by platelet endothelial cell adhesion molecule‐1 (PECAM‐1). We studied murine models of selective single‐dose (25 Gy) liver irradiation with and without TNF‐α application (2 μg/mouse; i.p.). In serum of wild‐type (wt)‐mice, irradiation induced a mild increase in hepatic damage marker aspartate aminotransferase (AST) in comparison to sham‐irradiated controls. AST levels further increased in mice treated with both irradiation and TNF‐α. Accordingly, elevated numbers of leucocytes and increased expression of the macrophage marker CD68 were observed in the liver of these mice. In parallel to hepatic damage, a consecutive decrease in expression of hepatic PECAM‐1 was found in mice that received radiation or TNF‐α treatment alone. The combination of radiation and TNF‐α induced an additional significant decline of PECAM‐1. Furthermore, increased expression of hepatic lipocalin‐2 (LCN‐2), a hepatoprotective protein, was detected at mRNA and protein levels after irradiation or TNF‐α treatment alone and the combination of both. Signal transducer and activator of transcription‐3 (STAT‐3) seems to be involved in the signalling cascade. To study the involvement of PECAM‐1 in hepatic damage more deeply, the liver of both wt‐ and PECAM‐1‐knock‐out‐mice were selectively irradiated (25 Gy). Thereby, ko‐mice showed higher liver damage as revealed by elevated AST levels, but also increased hepatoprotective LCN‐2 expression. Our studies show that TNF‐α has a pivotal role in radiation‐induced hepatic damage. It acts in concert with irradiation and its activity is modulated by PECAM‐1, which mediates pro‐ and anti‐inflammatory signalling.
Collapse
Affiliation(s)
- Gesa Malik
- Clinic for Gastroenterology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Jörg Wilting
- Department of Anatomy and Cell Biology, University Medical Center Göttingen, Göttingen, Germany
| | - Clemens Friedrich Hess
- Clinic for Radiotherapy and Radiooncology, University Medical Center Göttingen, Göttingen, Germany
| | - Giuliano Ramadori
- Clinic for Gastroenterology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Ihtzaz Ahmed Malik
- Clinic for Gastroenterology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany.,Department of Anatomy and Cell Biology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
14
|
Rampes S, Ma D. Hepatic ischemia-reperfusion injury in liver transplant setting: mechanisms and protective strategies. J Biomed Res 2019; 33:221-234. [PMID: 32383437 DOI: 10.7555/jbr.32.20180087] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatic ischemia-reperfusion injury is a major cause of liver transplant failure, and is of increasing significance due to increased use of expanded criteria livers for transplantation. This review summarizes the mechanisms and protective strategies for hepatic ischemia-reperfusion injury in the context of liver transplantation. Pharmacological therapies, the use of pre-and post-conditioning and machine perfusion are discussed as protective strategies. The use of machine perfusion offers significant potential in the reconditioning of liver grafts and the prevention of hepatic ischemia-reperfusion injury, and is an exciting and active area of research, which needs more study clinically.
Collapse
Affiliation(s)
- Sanketh Rampes
- Faculty of Life Sciences & Medicine, King's College London, London SE1 1U, UK
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, UK
| |
Collapse
|
15
|
Wu J, Zhao Y, Park YK, Lee JY, Gao L, Zhao J, Wang L. Loss of PDK4 switches the hepatic NF-κB/TNF pathway from pro-survival to pro-apoptosis. Hepatology 2018; 68:1111-1124. [PMID: 29603325 PMCID: PMC6165716 DOI: 10.1002/hep.29902] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 12/11/2022]
Abstract
It has been established that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) members promote survival by upregulating antiapoptotic genes and that genetic and pharmacological inhibition of NF-κB is required for tumor necrosis factor (TNF)-induced hepatocyte apoptosis. In this study, we demonstrate that this pro-survival pathway is switched to pro-apoptosis under pyruvate dehydrogenase kinase 4 (PDK4)-deficient conditions. PDK4-deficiency triggered hepatic apoptosis concomitantly with increased numbers of aberrant mitochondria, reactive oxygen species (ROS) production, sustained c-Jun N-terminal Kinase (JNK) activation, and reduction of glutathione (GSH). Interestingly, PDK4 retained p65 in cytoplasm via a direct protein-protein interaction. Disruption of PDK4-p65 association promoted p65 nuclear translocation. This, in turn, facilitated p65 binding to the TNF promoter to activate TNF-TNFR1 apoptotic pathway. Pdk4-/- livers were sensitized to Jo2 and D-(+)-Galactosamine /Lipopolysaccharide (GalN/LPS)-mediated apoptotic injury which was prevented by the inhibition of p65 or TNFR1. The pro-survival activity of TNF was shifted, which was switched to a pro-apoptotic activity in Pdk4-/- hepatocytes as a result of impaired activation of pro-survival NF-κB targets. Conclusion: PDK4 is indispensable to dictate the fate of TNF/NF-κB-mediated hepatocyte apoptosis. (Hepatology 2018).
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269,Corresponding author: Jianguo Wu (), 75 North Eagleville Rd., U3156, Storrs, CT 06269. Tel: 860-486-0857; Fax: 860-486-3303
| | - Yulan Zhao
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Young-Ki Park
- Department of Nutritional Sciences, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Ji-Young Lee
- Department of Nutritional Sciences, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, 250021, China,Institute of Endocrinology and metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, 250021, China,Institute of Endocrinology and metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Li Wang
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516,Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT 06520
| |
Collapse
|
16
|
Núñez K, Thevenot P, Alfadhli A, Cohen A. Complement Activation in Liver Transplantation: Role of Donor Macrosteatosis and Implications in Delayed Graft Function. Int J Mol Sci 2018; 19:ijms19061750. [PMID: 29899265 PMCID: PMC6032339 DOI: 10.3390/ijms19061750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 05/28/2018] [Accepted: 06/08/2018] [Indexed: 12/16/2022] Open
Abstract
The complement system anchors the innate inflammatory response by triggering both cell-mediated and antibody-mediated immune responses against pathogens. The complement system also plays a critical role in sterile tissue injury by responding to damage-associated molecular patterns. The degree and duration of complement activation may be a critical variable controlling the balance between regenerative and destructive inflammation following sterile injury. Recent studies in kidney transplantation suggest that aberrant complement activation may play a significant role in delayed graft function following transplantation, confirming results obtained from rodent models of renal ischemia/reperfusion (I/R) injury. Deactivating the complement cascade through targeting anaphylatoxins (C3a/C5a) might be an effective clinical strategy to dampen reperfusion injury and reduce delayed graft function in liver transplantation. Targeting the complement cascade may be critical in donor livers with mild to moderate steatosis, where elevated lipid burden amplifies stress responses and increases hepatocyte turnover. Steatosis-driven complement activation in the donor liver may also have implications in rejection and thrombolytic complications following transplantation. This review focuses on the roles of complement activation in liver I/R injury, strategies to target complement activation in liver I/R, and potential opportunities to translate these strategies to transplanting donor livers with mild to moderate steatosis.
Collapse
Affiliation(s)
- Kelley Núñez
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Paul Thevenot
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Abeer Alfadhli
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| | - Ari Cohen
- Institute of Translational Research, Ochsner Health System, New Orleans, LA 70121, USA.
| |
Collapse
|
17
|
nox2/cybb Deficiency Affects Zebrafish Retinotectal Connectivity. J Neurosci 2018; 38:5854-5871. [PMID: 29793976 DOI: 10.1523/jneurosci.1483-16.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 04/30/2018] [Accepted: 05/13/2018] [Indexed: 01/28/2023] Open
Abstract
NADPH oxidase (Nox)-derived reactive oxygen species (ROS) have been linked to neuronal polarity, axonal outgrowth, cerebellar development, regeneration of sensory axons, and neuroplasticity. However, the specific roles that individual Nox isoforms play during nervous system development in vivo remain unclear. To address this problem, we investigated the role of Nox activity in the development of retinotectal connections in zebrafish embryos. Zebrafish broadly express four nox genes (nox1, nox2/cybb, nox5, and duox) throughout the CNS during early development. Application of a pan-Nox inhibitor, celastrol, during the time of optic nerve (ON) outgrowth resulted in significant expansion of the ganglion cell layer (GCL), thinning of the ON, and a decrease in retinal axons reaching the optic tectum (OT). With the exception of GCL expansion, these effects were partially ameliorated by the addition of H2O2, a key ROS involved in Nox signaling. To address isoform-specific Nox functions, we used CRISPR/Cas9 to generate mutations in each zebrafish nox gene. We found that nox2/cybb chimeric mutants displayed ON thinning and decreased OT innervation. Furthermore, nox2/cybb homozygous mutants (nox2/cybb-/-) showed significant GCL expansion and mistargeted retinal axons in the OT. Neurite outgrowth from cultured zebrafish retinal ganglion cells was reduced by Nox inhibitors, suggesting a cell-autonomous role for Nox in these neurons. Collectively, our results show that Nox2/Cybb is important for retinotectal development in zebrafish.SIGNIFICANCE STATEMENT Most isoforms of NADPH oxidase (Nox) only produce reactive oxygen species (ROS) when activated by an upstream signal, making them ideal candidates for ROS signaling. Nox enzymes are present in neurons and their activity has been shown to be important for neuronal development and function largely by in vitro studies. However, whether Nox is involved in the development of axons and formation of neuronal connections in vivo has remained unclear. Using mutant zebrafish embryos, this study shows that a specific Nox isoform, Nox2/Cybb, is important for the establishment of axonal connections between retinal ganglion cells and the optic tectum.
Collapse
|
18
|
Hong JM, Lee SM. Heme oxygenase-1 protects liver against ischemia/reperfusion injury via phosphoglycerate mutase family member 5-mediated mitochondrial quality control. Life Sci 2018. [DOI: 10.1016/j.lfs.2018.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
19
|
Mortezaee K, Khanlarkhani N. Melatonin application in targeting oxidative‐induced liver injuries: A review. J Cell Physiol 2017; 233:4015-4032. [DOI: 10.1002/jcp.26209] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/04/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Keywan Mortezaee
- Department of AnatomySchool of MedicineKurdistan University of Medical SciencesSanandajIran
| | - Neda Khanlarkhani
- Department of Anatomy, School of MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
20
|
Bystrom P, Foley N, Toledo-Pereyra L, Quesnelle K. Ischemic preconditioning modulates ROS to confer protection in liver ischemia and reperfusion. EXCLI JOURNAL 2017; 16:483-496. [PMID: 28694752 PMCID: PMC5491905 DOI: 10.17179/excli2017-166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022]
Abstract
Ischemia reperfusion (IR) injury is a significant cause of morbidity and mortality in liver transplantation. When oxygen is reintroduced to the liver graft it initiates a cascade of molecular reactions leading to the release of reactive oxygen species (ROS) and pro-inflammatory cytokines. These soluble mediators propagate a sterile immune response to cause significant tissue damage. Ischemic preconditioning (IPC) is one method that reduces hepatocellular injury by altering the immune response and inhibiting the production of ROS. Studies quantifying the effects of IPC in humans have demonstrated an improved liver enzyme panel in patients receiving grafts pretreated with IPC as compared to patients receiving the standard of care. In our review, we explore current literature in the field in order to describe the mechanism through which IPC regulates the production of ROS and improves IR injury.
Collapse
Affiliation(s)
- Phillip Bystrom
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Biomedical Sciences
| | - Nicole Foley
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Biomedical Sciences
| | - Luis Toledo-Pereyra
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Surgery
| | - Kelly Quesnelle
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Biomedical Sciences
| |
Collapse
|
21
|
Hu C, Li L. Pre-conditions for eliminating mitochondrial dysfunction and maintaining liver function after hepatic ischaemia reperfusion. J Cell Mol Med 2017; 21:1719-1731. [PMID: 28301072 PMCID: PMC5571537 DOI: 10.1111/jcmm.13129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The liver, the largest organ with multiple synthesis and secretion functions in mammals, consists of hepatocytes and Kupffer, stem, endothelial, stellate and other parenchymal cells. Because of early and extensive contact with the external environment, hepatic ischaemia reperfusion (IR) may result in mitochondrial dysfunction, autophagy and apoptosis of cells and tissues under various pathological conditions. Because the liver requires a high oxygen supply to maintain normal detoxification and synthesis functions, it is extremely susceptible to ischaemia and subsequent reperfusion with blood. Consequently, hepatic IR leads to acute or chronic liver failure and significantly increases the total rate of morbidity and mortality through multiple regulatory mechanisms. An increasing number of studies indicate that mitochondrial structure and function are impaired after hepatic IR, but that the health of liver tissues or liver grafts can be effectively rescued by attenuation of mitochondrial dysfunction. In this review, we mainly focus on the subsequent therapeutic interventions related to the conservation of mitochondrial function involved in mitigating hepatic IR injury and the potential mechanisms of protection. Because mitochondria are abundant in liver tissue, clarification of the regulatory mechanisms between mitochondrial dysfunction and hepatic IR should shed light on clinical therapies for alleviating hepatic IR‐induced injury.
Collapse
Affiliation(s)
- Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Suyavaran A, Thirunavukkarasu C. Preconditioning methods in the management of hepatic ischemia reperfusion- induced injury: Update on molecular and future perspectives. Hepatol Res 2017; 47:31-48. [PMID: 26990696 DOI: 10.1111/hepr.12706] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 03/11/2016] [Indexed: 12/14/2022]
Abstract
Hepatic IR (ischemia reperfusion) injury is a commonly encountered obstacle in the post-operative management of hepatic surgery. Hepatic IR occurs during 'Pringle maneuver' for reduction of blood loss or during a brief period of cold storage followed by reperfusion of liver grafts. The stress induced during hepatic IR, triggers a spectrum of cellular responses leading to the varying degrees of hepatic complications which in turn affect the post operative care. Different preconditioning methods either activate or subdue different sets of molecular signals, resulting in varied levels of protection against hepatic IR injury. Yet, there is a serious lacuna in the knowledge regarding the choice of preconditioning methods and the resulting molecular changes in order to assess the efficiency and choice of these methods correctly. This review provides an update on the various preconditioning approaches such as surgical/ischemic, antioxidant, pharmaceutical and genetic preconditioning strategies published during last six years (2009-2015). Further, we discuss the attenuation or inhibition of specific inflammatory, apoptotic and necrotic markers in the various experimental models of liver IR subjected to different preconditioning strategies. While enlisting the controversies in the ischemic preconditioning strategy, we bring out the uncertainties in the existing molecular targets and their reliability in the attenuation of hepatic IR injury. Future research studies would include the novel preconditioning strategies employ i) the targeted gene silencing of key molecular targets inducing IR, ii) hyper expression of beneficial molecular signals against IR via gene transfer techniques. The above studies would see the combination of these latest techniques with the established preconditioning strategies for better post-operative hepatic management.
Collapse
Affiliation(s)
- Arumugam Suyavaran
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | |
Collapse
|
23
|
Wang J, Koh H, Zhou L, Bae U, Lee H, Bang IH, Ka S, Oh S, Bae EJ, Park B. Sirtuin 2 aggravates postischemic liver injury by deacetylating mitogen-activated protein kinase phosphatase-1. Hepatology 2017; 65:225-236. [PMID: 27532371 PMCID: PMC5215544 DOI: 10.1002/hep.28777] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022]
Abstract
UNLABELLED Sirtuin 2 (Sirt2) is known to negatively regulate anoxia-reoxygenation injury in myoblasts. Because protein levels of Sirt2 are increased in ischemia-reperfusion (I/R)-injured liver tissues, we examined whether Sirt2 is protective or detrimental against hepatic I/R injury. We overexpressed Sirt2 in the liver of C57BL/6 mice using a Sirt2 adenovirus. Wild-type and Sirt2 knockout mice were subjected to a partial (70%) hepatic ischemia for 45 minutes, followed by various periods of reperfusion. In another set of experiments, wild-type mice were pretreated intraperitoneally with AGK2, a Sirt2 inhibitor. Isolated hepatocytes and Kupffer cells from wild-type and Sirt2 knockout mice were subjected to hypoxia-reoxygenation injury to determine the in vitro effects of Sirt2. Mice subjected to I/R injury showed typical patterns of hepatocellular damage. Prior injection with Sirt2 adenovirus aggravated liver injury, as demonstrated by increases in serum aminotransferases, prothrombin time, proinflammatory cytokines, hepatocellular necrosis and apoptosis, and neutrophil infiltration relative to control virus-injected mice. Pretreatment with AGK2 resulted in significant improvements in serum aminotransferase levels and histopathologic findings. Similarly, experiments with Sirt2 knockout mice also revealed reduced hepatocellular injury. The molecular mechanism of Sirt2's involvement in this aggravation of hepatic I/R injury includes the deacetylation and inhibition of mitogen-activated protein kinase phosphatase-1 and consequent activation of mitogen-activated protein kinases. CONCLUSION Sirt2 is an aggravating factor during hepatic I/R injury. (Hepatology 2017;65:225-236).
Collapse
Affiliation(s)
- Jie Wang
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - Hyoung‐Won Koh
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - Lu Zhou
- Department of Sports MedicineTaishan Medical UniversityTaianShandongChina
| | - Ui‐Jin Bae
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - Hwa‐Suk Lee
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - In Hyuk Bang
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - Sun‐O Ka
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - Seon‐Hee Oh
- Department of Premedics, School of MedicineChosun UniversityGwangjuRepublic of Korea
| | - Eun Ju Bae
- College of PharmacyWoosuk UniversityWanjuJeonbukRepublic of Korea
| | - Byung‐Hyun Park
- Department of BiochemistryChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| |
Collapse
|
24
|
Landmann M, Sellmann C, Engstler AJ, Ziegenhardt D, Jung F, Brombach C, Bergheim I. Hops (Humulus lupulus) Content in Beer Modulates Effects of Beer on the Liver After Acute Ingestion in Female Mice. Alcohol Alcohol 2016; 52:48-55. [DOI: 10.1093/alcalc/agw060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 06/07/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023] Open
|
25
|
Hyperglycemia Aggravates Hepatic Ischemia Reperfusion Injury by Inducing Chronic Oxidative Stress and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3919627. [PMID: 27656261 PMCID: PMC5021880 DOI: 10.1155/2016/3919627] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 06/28/2016] [Accepted: 07/12/2016] [Indexed: 12/29/2022]
Abstract
Aim. To investigate whether hyperglycemia will aggravate hepatic ischemia reperfusion injury (HIRI) and the underlying mechanisms. Methods. Control and streptozotocin-induced diabetic Sprague-Dawley rats were subjected to partial hepatic ischemia reperfusion. Liver histology, transferase, inflammatory cytokines, and oxidative stress were assessed accordingly. Similarly, BRL-3A hepatocytes were subjected to hypoxia/reoxygenation (H/R) after high (25 mM) or low (5.5 mM) glucose culture. Cell viability, reactive oxygen species (ROS), and activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) were determined. Results. Compared with control, diabetic rats presented more severe hepatic injury and increased hepatic inflammatory cytokines and oxidative stress. HIRI in diabetic rats could be ameliorated by pretreatment of N-acetyl-L-cysteine (NAC) or apocynin. Excessive ROS generation and consequent Nrf2 and NF-κB translocation were determined after high glucose exposure. NF-κB translocation and its downstream cytokines were further increased in high glucose cultured group after H/R. While proper regulation of Nrf2 to its downstream antioxidases was observed in low glucose cultured group, no further induction of Nrf2 pathway by H/R after high glucose culture was identified. Conclusion. Hyperglycemia aggravates HIRI, which might be attributed to chronic oxidative stress and inflammation and potential malfunction of antioxidative system.
Collapse
|
26
|
Xu Q, Choksi S, Qu J, Jang J, Choe M, Banfi B, Engelhardt JF, Liu ZG. NADPH Oxidases Are Essential for Macrophage Differentiation. J Biol Chem 2016; 291:20030-41. [PMID: 27489105 DOI: 10.1074/jbc.m116.731216] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Indexed: 12/14/2022] Open
Abstract
NADPH oxidases (NOXs) are involved in inflammation, angiogenesis, tumor growth, and osteoclast differentiation. However, the role of NOX1 and NOX2 in macrophage differentiation and tumor progression is still elusive. Here we report that NOX1 and NOX2 are critical for the differentiation of monocytes to macrophages, the polarization of M2-type but not M1-type macrophages, and the occurrence of tumor-associated macrophages (TAMs). We found that deletion of both NOX1 and NOX2 led to a dramatic decrease in ROS production in macrophages and resulted in impaired efficiency in monocyte-to-macrophage differentiation and M2-type macrophage polarization. We further showed that NOX1 and NOX2 were critical for the activation of the MAPKs JNK and ERK during macrophage differentiation and that the deficiency of JNK and ERK activation was responsible for the failure of monocyte-to-macrophage differentiation, in turn affecting M2 macrophage polarization. Furthermore, we demonstrated that the decrease in M2 macrophages and TAMs, concomitant with the reduction of cytokine and chemokine secretion, contributed to the delay in wound healing and the inhibition of tumor growth and metastasis in NOX1/2 double knockout mice compared with WT mice. Collectively, these data provide direct evidence that NOX1 and NOX2 deficiency impairs macrophage differentiation and the occurrence of M2-type TAMs during tumor development.
Collapse
Affiliation(s)
- Qing Xu
- From the Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Swati Choksi
- From the Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Jianhui Qu
- From the Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Jonathan Jang
- From the Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Moran Choe
- From the Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Botond Banfi
- the Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242-1109
| | - John F Engelhardt
- the Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242-1109
| | - Zheng-Gang Liu
- From the Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| |
Collapse
|
27
|
Rebolledo RA, Hoeksma D, Hottenrott CMV, Bodar YJL, Ottens PJ, Wiersema-Buist J, Leuvenink HGD. Slow induction of brain death leads to decreased renal function and increased hepatic apoptosis in rats. J Transl Med 2016; 14:141. [PMID: 27193126 PMCID: PMC4872359 DOI: 10.1186/s12967-016-0890-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/29/2016] [Indexed: 11/17/2022] Open
Abstract
Background Donor brain death (BD) is an independent risk factor for graft survival in recipients. While in some patients BD results from a fast increase in intracranial pressure, usually associated with trauma, in others, intracranial pressure increases more slowly. The speed of intracranial pressure increase may be a possible risk factor for renal and hepatic graft dysfunction. This study aims to assess the effect of speed of BD induction on renal and hepatic injury markers. Methods BD induction was performed in 64 mechanically ventilated male Fisher rats by inflating a 4.0F Fogarty catheter in the epidural space. Rats were observed for 0.5, 1, 2 or 4 h following BD induction. Slow induction was achieved by inflating the balloon-catheter at a speed of 0.015 ml/min until confirmation of BD. Fast induction was achieved by inflating the balloon at 0.45 ml/min for 1 min. Plasma, kidney and liver tissue were collected for analysis. Results Slow BD induction led to higher plasma creatinine at all time points compared to fast induction. Furthermore, slow induction led to increased renal mRNA expression of IL-6, and renal MDA values after 4 h of BD compared to fast induction. Hepatic mRNA expression of TNF-α, Bax/Bcl-2, and protein expression of caspase-3 was significantly higher due to slow induction after 4 h of BD compared to fast induction. PMN infiltration was not different between fast and slow induction in both renal and hepatic tissue. Conclusion Slow induction of BD leads to poorer renal function compared to fast induction. Renal inflammatory and oxidative stress markers were increased. Liver function was not affected by speed of BD induction but hepatic inflammatory and apoptosis markers increased significantly due to slow induction compared to fast induction. These results provide initial proof that speed of BD induction influences detrimental renal and hepatic processes which could signify different donor management strategies for patients progressing to BD at different speeds.
Collapse
Affiliation(s)
- Rolando A Rebolledo
- Department of Surgery, University Medical Center Groningen, University of Groningen, CMC V, Y2144, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Physiopathology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Dane Hoeksma
- Department of Surgery, University Medical Center Groningen, University of Groningen, CMC V, Y2144, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Christina M V Hottenrott
- Department of Cardiothoracic Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Yves J L Bodar
- Department of Surgery, University Medical Center Groningen, University of Groningen, CMC V, Y2144, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Petra J Ottens
- Department of Surgery, University Medical Center Groningen, University of Groningen, CMC V, Y2144, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Janneka Wiersema-Buist
- Department of Surgery, University Medical Center Groningen, University of Groningen, CMC V, Y2144, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, University of Groningen, CMC V, Y2144, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
28
|
Li T, Guo B, Gao Y, Yu QH, Li JJ, Xian WJ, Jiang S, Zheng QC, Zhang Y. Tumor necrosis factor-alpha up-regulates expression of Jagged-1 and induces epithelial-mesenchymal transition in rat cholangiocytes in vitro. Shijie Huaren Xiaohua Zazhi 2016; 24:1806-1811. [DOI: 10.11569/wcjd.v24.i12.1806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of tumor necrosis factor-alpha (TNF-α) in the epithelial-mesenchymal transition (EMT) process in rat cholangiocytes in vitro.
METHODS: Primary rat cholangiocytes were treated with TNF-α (10 ng/mL) alone, TNF-α plus nuclear factor kappa B (NF-κB) inhibitor PDTC (50 µmol/L), or PDTC alone for 72 h. The expression of Jagged-1, mesenchymal markers [fibroblast-specific protein-1 (FSP-1), Vimentin and α-SMA] as well as epithelial marker CK19 was detected by Western blot. NF-κB binding activity was measured by EMSA. Migration ability and morphological changes of cholangiocytes were also examined.
RESULTS: In the TNF-α alone group, the protein levels of Jagged-1, FSP-1, Vimentin and α-SMA were up-regulated compared to control cells, whereas the expression of CK19 was down-regulated. The migration ability of cholangiocytes was increased and their shape changed from stone-like to fiber-like. For the TNF-α plus PTCD group and the PTCD alone group, no significant changes in EMT markers as well as migration ability were observed compared to control cells.
CONCLUSION: TNF-α is able to increase the expression of Jagged-1 and induce EMT in rat cholangiocytes in vitro possibly through activation of NF-κB signaling.
Collapse
|
29
|
Moreno-Càceres J, Mainez J, Mayoral R, Martín-Sanz P, Egea G, Fabregat I. Caveolin-1-dependent activation of the metalloprotease TACE/ADAM17 by TGF-β in hepatocytes requires activation of Src and the NADPH oxidase NOX1. FEBS J 2016; 283:1300-10. [PMID: 26815118 DOI: 10.1111/febs.13669] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/30/2015] [Accepted: 01/22/2016] [Indexed: 12/01/2022]
Abstract
Transforming growth factor-β (TGF-β) plays a dual role in hepatocytes, inducing both pro- and anti-apoptotic responses, the balance between which decides cell fate. Survival signals are mediated by the epidermal growth factor receptor (EGFR) pathway, which is activated by TGF-β. We have previously shown that caveolin-1 (CAV1) is required for activation of the metalloprotease tumour necrosis factor (TNF)-α-converting enzyme/a disintegrin and metalloproteinase 17 (TACE/ADAM17), and hence transactivation of the EGFR pathway. The specific mechanism by which TACE/ADAM17 is activated has not yet been determined. Here we show that TGF-β induces phosphorylation of sarcoma kinase (Src) in hepatocytes, a process that is impaired in Cav1(-/-) hepatocytes, coincident with a decrease in phosphorylated Src in detergent-resistant membrane fractions. TGF-β-induced activation of TACE/ADAM17 and EGFR phosphorylation were blocked using the Src inhibitor PP2. Cav1(+/+) hepatocytes showed early production of reactive oxygen species (ROS) induced by TGF-β, which was not seen in Cav1(-/-) cells. Production of ROS was inhibited by both the NADPH oxidase 1 (NOX1) inhibitor STK301831 and NOX1 knock-down, which also impaired TACE/ADAM17 activation and thus EGFR phosphorylation. Finally, neither STK301831 nor NOX1 silencing impaired Src phosphorylation, but PP2 blocked early ROS production, showing that Src is involved in NOX1 activation. As expected, inhibition of Src or NOX1 increased TGF-β-induced cell death in Cav1(+/+) cells. In conclusion, CAV1 is required for TGF-β-mediated activation of TACE/ADAM17 through a mechanism that involves phosphorylation of Src and NOX1-mediated ROS production.
Collapse
Affiliation(s)
| | - Jèssica Mainez
- Department of Cell Biology, Immunology and Neuroscience, School of Medicine, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Spain
| | - Rafael Mayoral
- Networked Biomedical Research Center on Hepatic and Digestive Diseases, Madrid, Spain.,Alberto Sols Biomedical Research Institute, CSIC-UAM, Madrid, Spain
| | - Paloma Martín-Sanz
- Networked Biomedical Research Center on Hepatic and Digestive Diseases, Madrid, Spain.,Alberto Sols Biomedical Research Institute, CSIC-UAM, Madrid, Spain
| | - Gustavo Egea
- Department of Cell Biology, Immunology and Neuroscience, School of Medicine, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Spain
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Physiological Sciences II, University of Barcelona, Spain
| |
Collapse
|
30
|
Molecular hydrogen attenuates hypoxia/reoxygenation injury of intrahepatic cholangiocytes by activating Nrf2 expression. Toxicol Lett 2015; 238:11-9. [PMID: 26276082 DOI: 10.1016/j.toxlet.2015.08.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/25/2015] [Accepted: 08/09/2015] [Indexed: 01/16/2023]
Abstract
Hypoxia/reoxygenation (H/R) injury of cholangiocytes causes serious biliary complications during hepatobiliary surgeries. Molecular hydrogen (H2) has been shown to be effective in protecting various cells and organs against oxidative stress injury. Human liver cholangiocytes were used to determine the potential protective effects of hydrogen against cholangiocyte H/R injury and explore the underlying mechanisms. We found that H2 ameliorated H/R-induced cholangiocytes apoptosis. Our study revealed that H2 activated NF-E2-related factor 2 (Nrf2) and downstream cytoprotective protein expression. However, the protective function of H2 was abolished when Nrf2 was silenced. Apoptosis in cholangiocytes isolated from a rat model of liver ischemia/reperfusion injury indicated that H2 significantly attenuates ischemia/reperfusion cholangiocyte injury in vivo. In conclusion, our study shows that H2 protects intrahepatic cholangiocytes from hypoxia/reoxygenation-induced apoptosis in vitro or in vivo, and this phenomenon may depend on activating Nrf2 expression.
Collapse
|
31
|
Zhou L, Koh HW, Bae UJ, Park BH. Aggravation of post-ischemic liver injury by overexpression of insulin-like growth factor binding protein 3. Sci Rep 2015; 5:11231. [PMID: 26073647 PMCID: PMC4466889 DOI: 10.1038/srep11231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/19/2015] [Indexed: 01/25/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is known to inhibit reperfusion-induced apoptosis. IGF-binding protein-3 (IGFBP-3) is the major circulating carrier protein for IGF-1 and induces apoptosis. In this study, we determined if IGFBP-3 was important in the hepatic response to I/R. To deliver IGFBP-3, we used an adenovirus containing IGFBP-3 cDNA (AdIGFBP-3) or an IGFBP-3 mutant devoid of IGF binding affinity but retaining IGFBP-3 receptor binding ability (AdIGFBP-3(GGG)). Mice subjected to I/R injury showed typical patterns of hepatocellular damage. Protein levels of IGFBP-3 were increased after reperfusion and showed a positive correlation with the extent of liver injury. Prior injection with AdIGFBP-3 aggravated liver injury: serum aminotransferases, prothrombin time, proinflammatory cytokines, hepatocellular necrosis and apoptosis, and neutrophil infiltration were markedly increased compared to control mice. A decrease in antioxidant potential and an upregulation of NADPH oxidase might have caused these aggravating effects of IGFBP-3. Experiments using HepG2 cells and N-acetylcysteine-pretreated mice showed a discernible effect of IGFBP-3 on reactive oxygen species generation. Lastly, AdIGFBP-3 abolished the beneficial effects of ischemic preconditioning and hypothermia. Mice treated with AdIGFBP-3(GGG) exhibited effects similar to those of AdIGFBP-3, suggesting a ligand-independent effect of IGFBP-3. Our results suggest IGFBP-3 as an aggravating factor during hepatic I/R injury.
Collapse
Affiliation(s)
- Lu Zhou
- 1] Department of Sports Medicine, Taishan Medical University, Taian, Shandong, 271-000, China [2] Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk, 561-756, Republic of Korea
| | - Hyoung-Won Koh
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk, 561-756, Republic of Korea
| | - Ui-Jin Bae
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk, 561-756, Republic of Korea
| | - Byung-Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk, 561-756, Republic of Korea
| |
Collapse
|
32
|
Idh1 protects murine hepatocytes from endotoxin-induced oxidative stress by regulating the intracellular NADP(+)/NADPH ratio. Cell Death Differ 2015; 22:1837-45. [PMID: 25882048 PMCID: PMC4648331 DOI: 10.1038/cdd.2015.38] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 01/11/2023] Open
Abstract
Isocitrate dehydrogenase-1 (Idh1) is an important metabolic enzyme that produces NADPH by converting isocitrate to α-ketoglutarate. Idh1 is known to reduce reactive oxygen species (ROS) induced in cells by treatment with lipopolysaccharide (LPS) in vitro. Here, we used Idh1-deficient knockout (Idh1 KO) mice to investigate the role of Idh1 in antioxidant defense in vivo. Idh1 KO mice showed heightened susceptibility to death induced by LPS and exhibited increased serum levels of inflammatory cytokines such as tumor necrosis factor-α and interleukin-6. The serum of LPS-injected Idh1 KO mice also contained elevated levels of AST, a marker of inflammatory liver damage. Furthermore, after LPS injection, livers of Idh1 KO mice showed histological evidence of elevated oxidative DNA damage compared with livers of wild-type (WT) mice. Idh1 KO livers showed a faster and more pronounced oxidative stress than WT livers. In line with that, Idh1 KO hepatocytes showed higher ROS levels and an increase in the NADP(+)/NADPH ratio when compared with hepatocytes isolated from WT mice. These results suggest that Idh1 has a physiological function in protecting cells from oxidative stress by regulating the intracellular NADP(+)/NADPH ratio. Our findings suggest that stimulation of Idh1 activity may be an effective therapeutic strategy for reducing oxidative stress during inflammatory responses, including the early stages of septic shock.
Collapse
|
33
|
Sun M, Cui W, Woody SK, Staudinger JL. Pregnane X receptor modulates the inflammatory response in primary cultures of hepatocytes. Drug Metab Dispos 2014; 43:335-43. [PMID: 25527709 DOI: 10.1124/dmd.114.062307] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bacterial sepsis is characterized by a rapid increase in the expression of inflammatory mediators to initiate the acute phase response in liver. Inflammatory mediator release is counterbalanced by the coordinated expression of anti-inflammatory molecules such as interleukin 1 receptor antagonist (IL1-Ra) through time. This study determined whether activation of pregnane X receptor (PXR, NR1I2) alters the lipopolysaccharide (LPS)-inducible gene expression program in primary cultures of hepatocytes (PCHs). Preactivation of PXR for 24 hours in PCHs isolated from wild-type mice suppressed the subsequent LPS-inducible expression of the key inflammatory mediators interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor α (TNFα) but not in PCHs isolated from Pxr-null (PXR-knockout [KO]) mice. Basal expression of key inflammatory cytokines was elevated in PCHs from PXR-KO mice. Stimulation of PCHs from PXR-KO mice with LPS alone produced enhanced levels of IL-1β when compared with wild-type mice. Experiments performed using PCHs from both humanized-PXR transgenic mice as well as human donors indicate that prolonged activation of PXR produces an increased secretion of IL1-Ra from cells through time. Our data reveal a working model that describes a pivotal role for PXR in both inhibiting as well as in resolving the inflammatory response in hepatocytes. Understanding the molecular details of how PXR is converted from a positive regulator of drug-metabolizing enzymes into a transcriptional suppressor of inflammation in liver will provide new pharmacologic strategies for modulating inflammatory-related diseases in the liver and intestine.
Collapse
Affiliation(s)
- Mengxi Sun
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas
| | - Wenqi Cui
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas
| | - Sarah K Woody
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas
| | - Jeff L Staudinger
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas
| |
Collapse
|
34
|
Superoxide deficiency attenuates promotion of hepatocarcinogenesis by cytotoxicity in NADPH oxidase knockout mice. Arch Toxicol 2014; 89:1383-93. [DOI: 10.1007/s00204-014-1298-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/17/2014] [Indexed: 01/26/2023]
|
35
|
Choi J, Corder NLB, Koduru B, Wang Y. Oxidative stress and hepatic Nox proteins in chronic hepatitis C and hepatocellular carcinoma. Free Radic Biol Med 2014; 72:267-84. [PMID: 24816297 PMCID: PMC4099059 DOI: 10.1016/j.freeradbiomed.2014.04.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 04/16/2014] [Accepted: 04/18/2014] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common liver cancer and a leading cause of cancer-related mortality in the world. Hepatitis C virus (HCV) is a major etiologic agent of HCC. A majority of HCV infections lead to chronic infection that can progress to cirrhosis and, eventually, HCC and liver failure. A common pathogenic feature present in HCV infection, and other conditions leading to HCC, is oxidative stress. HCV directly increases superoxide and H2O2 formation in hepatocytes by elevating Nox protein expression and sensitizing mitochondria to reactive oxygen species generation while decreasing glutathione. Nitric oxide synthesis and hepatic iron are also elevated. Furthermore, activation of phagocytic NADPH oxidase (Nox) 2 of host immune cells is likely to exacerbate oxidative stress in HCV-infected patients. Key mechanisms of HCC include genome instability, epigenetic regulation, inflammation with chronic tissue injury and sustained cell proliferation, and modulation of cell growth and death. Oxidative stress, or Nox proteins, plays various roles in these mechanisms. Nox proteins also function in hepatic fibrosis, which commonly precedes HCC, and Nox4 elevation by HCV is mediated by transforming growth factor β. This review summarizes mechanisms of oncogenesis by HCV, highlighting the roles of oxidative stress and hepatic Nox enzymes in HCC.
Collapse
Affiliation(s)
- Jinah Choi
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA.
| | - Nicole L B Corder
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA
| | - Bhargav Koduru
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA
| | - Yiyan Wang
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA
| |
Collapse
|
36
|
Effect of acute beer ingestion on the liver: studies in female mice. Eur J Nutr 2014; 54:465-74. [DOI: 10.1007/s00394-014-0730-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 06/03/2014] [Indexed: 12/20/2022]
|
37
|
Hartmann P, Fet N, Garab D, Szabó A, Kaszaki J, Srinivasan PK, Tolba RH, Boros M. L-alpha-glycerylphosphorylcholine reduces the microcirculatory dysfunction and nicotinamide adenine dinucleotide phosphate-oxidase type 4 induction after partial hepatic ischemia in rats. J Surg Res 2014; 189:32-40. [DOI: 10.1016/j.jss.2013.12.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/21/2013] [Accepted: 12/30/2013] [Indexed: 10/25/2022]
|
38
|
Garab D, Fet N, Szabó A, Tolba RH, Boros M, Hartmann P. Remote ischemic preconditioning differentially affects NADPH oxidase isoforms during hepatic ischemia–reperfusion. Life Sci 2014; 105:14-21. [DOI: 10.1016/j.lfs.2014.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/07/2014] [Indexed: 01/21/2023]
|
39
|
Spencer NY, Engelhardt JF. The basic biology of redoxosomes in cytokine-mediated signal transduction and implications for disease-specific therapies. Biochemistry 2014; 53:1551-64. [PMID: 24555469 PMCID: PMC3985689 DOI: 10.1021/bi401719r] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Redox
reactions have been established as major biological players
in many cellular signaling pathways. Here we review mechanisms of
redox signaling with an emphasis on redox-active signaling endosomes.
Signals are transduced by relatively few reactive oxygen species (ROS),
through very specific redox modifications of numerous proteins and
enzymes. Although ROS signals are typically associated with cellular
injury, these signaling pathways are also critical for maintaining
cellular health at homeostasis. An important component of ROS signaling
pertains to localization and tightly regulated signal transduction
events within discrete microenvironments of the cell. One major aspect
of this specificity is ROS compartmentalization within membrane-enclosed
organelles such as redoxosomes (redox-active endosomes) and the nuclear
envelope. Among the cellular proteins that produce superoxide are
the NADPH oxidases (NOXes), transmembrane proteins that are implicated
in many types of redox signaling. NOXes produce superoxide on only
one side of a lipid bilayer; as such, their orientation dictates the
compartmentalization of ROS and the local control of signaling events
limited by ROS diffusion and/or movement through channels associated
with the signaling membrane. NOX-dependent ROS signaling pathways
can also be self-regulating, with molecular redox sensors that limit
the local production of ROS required for effective signaling. ROS
regulation of the Rac-GTPase, a required co-activator of many NOXes,
is an example of this type of sensor. A deeper understanding of redox
signaling pathways and the mechanisms that control their specificity
will provide unique therapeutic opportunities for aging, cancer, ischemia-reperfusion
injury, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Netanya Y Spencer
- Department of Anatomy and Cell Biology, The University of Iowa , Iowa City, Iowa 52242-1009, United States
| | | |
Collapse
|
40
|
Yun N, Cho HI, Lee SM. Impaired autophagy contributes to hepatocellular damage during ischemia/reperfusion: heme oxygenase-1 as a possible regulator. Free Radic Biol Med 2014; 68:168-77. [PMID: 24365205 DOI: 10.1016/j.freeradbiomed.2013.12.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 11/15/2013] [Accepted: 12/11/2013] [Indexed: 12/14/2022]
Abstract
Liver ischemia and reperfusion (I/R) injury is characterized by oxidative stress that is accompanied by alterations of the endogenous defensive system. Emerging evidence suggests a protective role for autophagy induced by multiple stressors including reactive oxygen species. Meanwhile, heme oxygenase-1 (HO-1) has long been implicated in cytoprotection against oxidative stress in vitro and in vivo. Therefore, we investigated the impact of autophagy in the pathogenesis of liver I/R and its molecular mechanisms, particularly its linkage to HO-1. By using transmission electron microscopic analysis and biochemical autophagic flux assays with microtubule-associated protein 1 light chain 3-II, and beclin-1, representative autophagy markers, and p62, a selective substrate for autophagy, we found that reperfusion reduced autophagy both in the rat liver and in primary cultured hepatocytes. When autophagy was further inhibited with chloroquine or wortmannin, I/R-induced hepatocellular injury was aggravated. While livers that underwent I/R showed increased levels of mammalian target of rapamaycin and calpain 1 and 2, inhibition of calpain 1 and 2 induced an autophagic response in hepatocytes subjected to hypoxia/reoxygenation. HO-1 increased autophagy, and HO-1 reduced I/R-induced calcium overload in hepatocytes and prevented calpain 2 activation both in vivo and in vitro. Taken together, these findings suggest that the impaired autophagy during liver I/R, which is mediated by calcium overload and calpain activation, contributes to hepatocellular damage and the HO-1 system protects the liver from I/R injury through enhancing autophagy.
Collapse
Affiliation(s)
- Nari Yun
- School of Pharmacy, Sungkyunkwan University, 300 Cheoncheon-dong, Jangan-gu, Suwon, Gyeonggi-do 440-746, Republic of Korea
| | - Hong-Ik Cho
- School of Pharmacy, Sungkyunkwan University, 300 Cheoncheon-dong, Jangan-gu, Suwon, Gyeonggi-do 440-746, Republic of Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, 300 Cheoncheon-dong, Jangan-gu, Suwon, Gyeonggi-do 440-746, Republic of Korea.
| |
Collapse
|
41
|
PTPRO plays a dual role in hepatic ischemia reperfusion injury through feedback activation of NF-κB. J Hepatol 2014; 60:306-12. [PMID: 24128416 DOI: 10.1016/j.jhep.2013.09.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/15/2013] [Accepted: 09/30/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Nuclear factor-κB (NF-κB) activation in hepatocytes and macrophages appeared as a double-edged-sword in hepatic ischemia reperfusion (IR) injury. Protein tyrosine phosphatase receptor type O (PTPRO) was recently identified as a potential activator of c-Src, which can in turn activate the NF-κB pathway. In this study, we aimed to determine the change and function of PTPRO in hepatocytes and macrophages during IR. METHODS Clinical patients with benign liver condition undergoing liver surgery were recruited in our study. Wild type (WT) and ptpro(-/-) C57BL/6 mice were processed to construct hepatic IR models. Isolated mouse hepatocytes and macrophages were treated with peroxide or TNFα in vitro. RESULTS In human and mouse IR models, PTPRO level was decreased in the early phase but reversed in the late phase. In vitro studies demonstrated that NF-κB up-regulated PTPRO transcription. Using ptpro(-/-) mice and primary cells, we found that PTPRO deficiency resulted in reduction of NF-κB activation in both hepatocytes and macrophages and was correlated to c-Src phosphorylation; PTPRO in hepatocytes alleviated, but PTPROt in macrophages exacerbated IR injury. CONCLUSIONS PTPRO activates NF-κB in a positive feedback manner, and plays a dual role in hepatic IR injury.
Collapse
|